At2g35430 Antibody

Shipped with Ice Packs
In Stock

Description

What is the At2g35430 Antibody?

The At2g35430 antibody is a polyclonal antibody raised against the protein product of the At2g35430 gene in Arabidopsis thaliana. This gene’s protein product remains under investigation, but antibodies like this enable researchers to study its expression patterns, interactions, and roles in plant physiology .

Potential Research Applications

While direct studies on At2g35430 are not detailed in the provided sources, analogous plant antibodies are typically employed in:

  • Protein Localization: Tracking spatial expression in plant tissues via immunohistochemistry.

  • Gene Knockout Validation: Confirming the absence of the target protein in mutant lines.

  • Interaction Studies: Identifying binding partners through co-immunoprecipitation .

Functional Insights

  • Stress responses (e.g., drought, salinity).

  • Developmental regulation.

  • Metabolic processes .

Limitations and Future Directions

Current data gaps include:

  • Protein Function: No direct studies on At2g35430’s biological role.

  • Validation Metrics: Lack of detailed specificity or cross-reactivity data.
    Future research could pair this antibody with transcriptomic or proteomic workflows to clarify its target’s function .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
At2g35430 antibody; T32F12.19Zinc finger CCCH domain-containing protein 28 antibody; AtC3H28 antibody
Target Names
At2g35430
Uniprot No.

Q&A

What is At2g35430 and why is it significant in Arabidopsis thaliana research?

At2g35430 refers to a specific gene locus in Arabidopsis thaliana (Mouse-ear cress), a widely used model organism in plant molecular biology. This gene encodes a protein with Uniprot identifier Q5PP65. Researchers study this protein to understand plant development, stress responses, and cellular signaling pathways. The antibody against At2g35430 enables detection and quantification of the protein in various experimental contexts, facilitating functional studies of gene expression, protein localization, and interaction networks. Understanding At2g35430's role contributes to fundamental knowledge about plant biology and potentially informs agricultural applications for crop improvement and stress resistance.

What controls should be incorporated when working with At2g35430 antibody?

Control TypePurposeImplementation
Positive ControlValidates antibody activitySamples known to express At2g35430 (wild-type A. thaliana tissue)
Negative ControlAssesses non-specific bindingSamples lacking target (either knockout lines or non-plant tissue)
Loading ControlNormalizes protein amountsAnti-tubulin or anti-actin antibodies for Western blots
Secondary Antibody ControlEvaluates secondary antibody specificityPrimary antibody omitted, secondary antibody only
Isotype ControlMeasures background from antibody classNon-specific rabbit IgG matched to concentration
Blocking PeptideConfirms epitope specificityPre-incubate antibody with immunogen peptide

Incorporating these controls helps distinguish true signals from artifacts and enables accurate data interpretation. For Western blots, molecular weight markers confirm target identity, while for immunohistochemistry, parallel sections without primary antibody establish background staining levels. Standardizing control protocols across experiments improves reproducibility and enables meaningful cross-experimental comparisons. Document all control results meticulously in laboratory notebooks for future reference and publication requirements.

How can researchers assess At2g35430 antibody specificity?

Assessing antibody specificity is crucial for result interpretation. Start with Western blot analysis using wild-type Arabidopsis thaliana tissue to confirm the antibody detects a band of the expected molecular weight. Compare this with tissue from At2g35430 knockout or knockdown lines, where the signal should be absent or significantly reduced. Peptide competition assays, where the antibody is pre-incubated with excess immunizing peptide before application to samples, should abolish specific binding if the antibody is truly specific. For immunoprecipitation applications, mass spectrometry analysis of pulled-down proteins can validate target specificity. Cross-reactivity testing against related proteins helps establish detection boundaries. When available, comparing results from different antibodies targeting the same protein but recognizing different epitopes provides additional validation. These approaches collectively build confidence in antibody specificity before proceeding to experimental applications.

What strategies can improve Western blot sensitivity when using At2g35430 antibody?

Enhancing Western blot sensitivity for At2g35430 detection requires systematic optimization at multiple steps. Begin by testing different protein extraction buffers that preserve the native state of plant proteins while minimizing protease activity. For Arabidopsis thaliana tissues, a buffer containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% Triton X-100, 0.5% sodium deoxycholate, and protease inhibitor cocktail often yields good results. Experiment with loading higher protein amounts (50-100 μg) when target expression is expected to be low. Consider using gradient gels (4-20%) to improve separation and transfer efficiency. For membrane transfer, semi-dry systems may be suboptimal for plant proteins; instead, wet transfer at 30V overnight at 4°C often improves transfer efficiency of recalcitrant proteins.

High-sensitivity ECL substrates containing enhanced luminol derivatives can increase detection limits by 10-50 fold compared to standard ECL. Signal amplification systems like tyramide signal amplification or biotinyl tyramide amplification can further enhance sensitivity by 10-200 fold, though these require careful optimization to prevent background increase. Digital imaging systems with cooled CCD cameras offer superior sensitivity compared to film, capturing a broader dynamic range and enabling more precise quantification. Finally, switching from PVDF to nitrocellulose membranes sometimes improves signal-to-noise ratio for specific antibodies, though this parameter needs empirical testing for each antibody-antigen pair.

How can researchers validate At2g35430 antibody specificity in genetic knockout studies?

Genetic knockout validation represents the gold standard for antibody specificity confirmation. For At2g35430, researchers should ideally obtain or generate T-DNA insertion lines, CRISPR/Cas9 knockout lines, or RNAi knockdown lines targeting this gene. When performing Western blot analysis, wild-type plants should show a distinct band at the expected molecular weight, while the signal should be absent (knockout) or significantly reduced (knockdown) in the mutant lines. Including heterozygous plants can demonstrate dose-dependent signal reduction, further confirming specificity.

Complementation experiments, where the At2g35430 gene is reintroduced into knockout lines under a suitable promoter, should restore the antibody signal, providing additional validation. For proteins with tissue-specific expression, comparing antibody signals between tissues known to express or lack At2g35430 provides another validation approach. Quantitative PCR measuring At2g35430 transcript levels should correlate with protein levels detected by the antibody across different tissues or conditions, though post-transcriptional regulation can sometimes disrupt this correlation. When knockout is lethal, inducible expression systems or tissue-specific knockouts provide alternative validation approaches. Document all validation experiments thoroughly with appropriate controls for publication and laboratory records.

What troubleshooting approaches are recommended when At2g35430 antibody yields inconsistent results?

When facing inconsistent results with At2g35430 antibody, implement a systematic troubleshooting approach. First, verify antibody integrity by checking storage conditions, freeze-thaw history, and expiration date. Prepare fresh working dilutions from primary stocks if needed. Next, examine protein extraction methods, as plant tissues contain polyphenols and other compounds that can interfere with immunodetection. Using polyvinylpolypyrrolidone (PVPP) in extraction buffers can adsorb these interfering compounds. Increase the concentration of reducing agents like DTT (up to 10 mM) to ensure complete protein denaturation.

For Western blots showing variable results, optimize blocking conditions by testing different blockers (5% non-fat milk, 5% BSA, commercial blocking solutions) as plant proteins may interact differently with these agents. Adjust primary antibody concentration and incubation conditions (1:500-1:5000 dilution range, 4°C overnight vs. room temperature for 1-2 hours). For ELISA applications, check for edge effects in plates and implement temperature equilibration steps before readings. Consider batch effects by using the same antibody lot across comparative experiments. Document all parameters meticulously, including sample preparation details, buffer compositions, and incubation times/temperatures. When persistent issues occur, perform parallel experiments with multiple antibody lots or alternative detection methods to distinguish between antibody-specific and method-specific variability sources.

How does post-translational modification affect At2g35430 antibody binding and detection?

Post-translational modifications (PTMs) can significantly impact antibody binding to At2g35430 protein. Phosphorylation, a common regulatory PTM in plants, may either mask or create epitopes, leading to false negative or positive results. To assess this impact, researchers should compare dephosphorylated samples (treated with lambda phosphatase) with untreated samples in Western blots. If signal intensity changes after phosphatase treatment, the antibody likely recognizes a phosphorylation-dependent epitope. Similarly, other PTMs like ubiquitination, sumoylation, or glycosylation can affect antibody recognition.

For comprehensive analysis, two-dimensional gel electrophoresis followed by Western blotting can separate protein isoforms with different PTMs, revealing which forms the antibody detects. Mass spectrometry analysis of immunoprecipitated At2g35430 can identify specific modification sites. Researchers should also be aware that stress conditions, developmental stages, or plant hormones may alter the PTM pattern of At2g35430, potentially affecting antibody detection across experimental conditions. When investigating protein function, using multiple antibodies recognizing different epitopes can help distinguish between protein absence and epitope masking due to PTMs. This comprehensive approach enables more accurate interpretation of results, particularly when studying dynamic cellular processes where protein modifications play regulatory roles.

What methodologies combine At2g35430 antibody with other techniques for comprehensive functional studies?

Integrating multiple techniques with At2g35430 antibody enables comprehensive functional characterization. Chromatin immunoprecipitation (ChIP) coupled with sequencing (ChIP-seq) can identify DNA binding sites if At2g35430 functions as a transcription factor or chromatin-associated protein. Co-immunoprecipitation (Co-IP) followed by mass spectrometry reveals protein interaction partners, building protein-protein interaction networks. For subcellular localization, combine immunofluorescence microscopy with organelle-specific markers to precisely map protein distribution.

Proximity-dependent biotin identification (BioID) or proximity ligation assay (PLA) with At2g35430 antibody can identify transient or weak interactions often missed by traditional Co-IP. For temporal dynamics, combine antibody detection with real-time measurements using techniques like fluorescence resonance energy transfer (FRET) or fluorescence recovery after photobleaching (FRAP) using fluorescently tagged versions of At2g35430. Multiplexed immunoassays allow simultaneous quantification of At2g35430 alongside other proteins of interest.

Integrated MethodologyApplicationKey Advantage
ChIP-seqDNA binding site identificationGenome-wide binding profile
Co-IP/Mass SpectrometryProtein interaction mappingUnbiased partner identification
Immunofluorescence/Confocal MicroscopySubcellular localizationSpatial distribution in native context
BioID/PLAWeak/transient interaction detectionCaptures interactions in living cells
FRAP/FRETProtein dynamicsReal-time kinetic measurements
Ribosome profiling with Western blotTranslation efficiency correlationLinks transcription to protein synthesis

These integrated approaches provide multidimensional insights into At2g35430 function, connecting molecular interactions with cellular processes and organismal phenotypes.

What are the optimal blocking conditions for reducing background when using At2g35430 antibody?

Background reduction requires systematic optimization of blocking conditions. While the At2g35430 antibody is supplied in a buffer containing 50% glycerol, 0.01M PBS (pH 7.4), and 0.03% Proclin 300 , blocking protocols must be empirically determined. For Western blot applications, comparing 5% non-fat dry milk, 3-5% BSA, and commercial blockers (like SuperBlock or Odyssey Blocking Buffer) often reveals significant differences in signal-to-noise ratios. Plant samples typically contain endogenous biotin and other compounds that can increase background, making casein-based blockers sometimes preferable to milk-based options.

The blocking duration significantly impacts background; test both standard (1 hour at room temperature) and extended protocols (overnight at 4°C). For immunohistochemistry applications, adding 0.1-0.3% Triton X-100 to blocking solutions improves tissue penetration while reducing non-specific binding. When using fluorescent secondary antibodies, auto-fluorescence from plant tissues must be addressed through additional blocking steps with 0.1% sodium borohydride or commercial auto-fluorescence quenchers. Document comparative results methodically, as optimal blocking conditions may vary between experimental contexts and tissue types. Regardless of the chosen method, maintain consistency in blocking protocols across comparative experiments to ensure reliable analysis.

How should researchers optimize protein extraction for At2g35430 detection in different plant tissues?

Protein extraction optimization is crucial for reliable At2g35430 detection across various plant tissues, which differ in composition and interfering compounds. For Arabidopsis leaf tissue, a buffer containing 50 mM HEPES-KOH (pH 7.5), 10 mM MgCl₂, 1 mM EDTA, 10% glycerol, 0.1% Triton X-100, 1 mM PMSF, and protease inhibitor cocktail typically yields good results. For root tissues, which contain higher levels of phenolic compounds, adding 2% polyvinylpolypyrrolidone (PVPP) and increasing DTT concentration to 5-10 mM improves extraction quality.

When extracting from siliques or seeds, which contain high levels of storage proteins and oils, a more stringent buffer containing 7M urea, 2M thiourea, 4% CHAPS, 20 mM DTT, and 1% plant protease inhibitor cocktail may be necessary. Sample grinding methodology also impacts extraction efficiency; cryogenic grinding with liquid nitrogen preserves protein integrity better than room temperature extraction. For recalcitrant tissues, consider sonication (3-5 short pulses at 30% amplitude) following initial extraction to improve protein release. Centrifugation speeds and durations should be optimized; higher speeds (14,000-16,000 × g) for longer durations (15-20 minutes) often yield cleaner supernatants for immunodetection. Always quantify protein concentration after extraction using Bradford or BCA assays, and normalize loading amounts carefully for comparative analyses across different tissues or conditions.

What strategies can researchers employ to validate At2g35430 antibody for immunoprecipitation?

Validating At2g35430 antibody for immunoprecipitation (IP) requires a systematic approach to ensure both efficient target capture and minimal non-specific binding. Begin with a pilot IP experiment comparing the antibody to normal rabbit IgG as a negative control. Input, unbound, and eluted fractions should be analyzed by Western blot to confirm enrichment of At2g35430 in the eluted fraction. Cross-linking the antibody to protein A/G beads using dimethyl pimelimidate (DMP) can prevent antibody co-elution, which sometimes masks results.

For stringent validation, perform parallel IPs from wild-type and At2g35430 knockdown/knockout tissues. The target band should show reduced or absent signal in the mutant IP elution. To optimize IP conditions, test different lysis buffers varying in detergent composition (Triton X-100, NP-40, or CHAPS) and salt concentration (150-500 mM NaCl). Higher stringency wash buffers containing 0.1-0.5% SDS or 0.1% sodium deoxycholate can reduce non-specific binding, though this must be balanced against potential loss of specific interactions.

Mass spectrometry analysis of IP eluates provides unbiased validation, ideally showing At2g35430 as the most abundant protein in the experimental sample compared to the IgG control. For challenging targets, employing epitope-tagged versions of At2g35430 (in parallel with the native protein) allows comparison of IP efficiency. Finally, successful IP conditions should be documented in detailed protocols specifying buffer compositions, incubation times and temperatures, wash stringency, and elution conditions to ensure reproducibility across experiments.

How can researchers quantitatively compare At2g35430 expression across different experimental conditions?

Quantitative comparison of At2g35430 expression requires careful experimental design and analytical approaches. For Western blot-based quantification, always include a standard curve using recombinant protein or a pooled reference sample at multiple dilutions to ensure measurements fall within the linear detection range. Digital imaging systems with 16-bit dynamic range capture provide superior quantification compared to film-based methods. Normalize target signals to validated housekeeping proteins (e.g., actin, tubulin, or GAPDH) chosen based on expression stability under your experimental conditions.

For more precise quantification, consider multiplex fluorescent Western blotting, where the target and reference proteins are simultaneously detected with spectrally distinct fluorophores, eliminating stripping and reprobing variability. ELISA provides an alternative quantification method with potentially higher throughput, but requires careful validation of antibody performance in this format. When analyzing multiple samples, minimize batch effects by processing all comparative samples simultaneously on the same membrane or plate.

Statistical analysis should account for technical replicates (multiple measurements from the same biological sample) and biological replicates (independent experimental units). Appropriate statistical tests depend on experimental design; for comparing multiple conditions, ANOVA followed by post-hoc tests is typically appropriate, while t-tests are suitable for binary comparisons. Report both fold changes and absolute expression levels when possible. For time-course experiments, consider area-under-curve analyses rather than point-by-point comparisons. Comprehensive documentation of all normalization methods, technical parameters, and statistical approaches is essential for result interpretation and reproducibility.

What are the recommended approaches for multiplexing At2g35430 antibody with other antibodies?

Multiplexing At2g35430 antibody with other antibodies enables simultaneous analysis of multiple proteins, providing insights into co-regulation and interaction networks. For immunofluorescence applications, select secondary antibodies with spectrally distinct fluorophores that minimize channel overlap. When using the rabbit polyclonal At2g35430 antibody , pair it with antibodies raised in different species (mouse, goat, or chicken) to enable clean discrimination through species-specific secondary antibodies.

For Western blot multiplexing, fluorescent detection systems offer advantages over chemiluminescence, allowing simultaneous imaging of multiple targets without stripping and reprobing. If sequential detection is necessary due to similar molecular weights of targets, document complete stripping using secondary antibody-only controls before reprobing. When both targets are detected with rabbit antibodies, size separation (>15 kDa difference) is essential for reliable discrimination.

Multiplexing ApproachApplicationConsiderations
Fluorescent Western blotQuantitative protein co-expressionRequires specialized imaging systems
Multi-color immunofluorescenceProtein co-localizationNeeds careful spectral separation
Sequential chromogenic IHCTissue co-expression patternsTime-intensive but accessible equipment
Mass cytometry (CyTOF)Single-cell protein networksRequires metal-conjugated antibodies
Proximity ligation assayProtein-protein interactionsDetects only closely associated proteins

For each multiplexing approach, optimization should include antibody titration in the multiplex context, as optimal concentrations often differ from single-detection protocols. Cross-reactivity testing is essential, particularly when using multiple polyclonal antibodies. Well-designed multiplexing controls should include single-antibody staining for each target to verify signal specificity and localization in the multiplex context.

How can At2g35430 antibody be utilized in plant stress response studies?

The At2g35430 antibody offers valuable insights into plant stress response mechanisms when applied within appropriate experimental frameworks. For studying abiotic stresses (drought, salt, temperature extremes), researchers should collect tissue samples at multiple timepoints following stress application, extracting proteins under denaturing conditions to capture stress-induced protein modifications. Western blot analysis using the At2g35430 antibody can reveal changes in protein abundance, while additional techniques like Phos-tag SDS-PAGE can detect stress-induced phosphorylation events that may alter protein function.

Subcellular fractionation followed by immunoblotting can identify stress-induced translocation of At2g35430 between cellular compartments, potentially indicating functional changes. For in situ localization studies, compare At2g35430 distribution patterns in control versus stressed tissues using immunohistochemistry, noting any changes in tissue-specific expression or subcellular localization. Combining these approaches with transcriptomic data allows correlation between transcript and protein levels during stress responses, potentially revealing post-transcriptional regulation mechanisms.

Co-immunoprecipitation under different stress conditions can identify stress-specific protein interaction partners, building dynamic interaction networks. When using these approaches, careful experimental design should include appropriate stress intensity and duration, with time-course sampling to capture both early signaling events and later acclimation responses. Parallel analysis of known stress markers provides important context for interpreting At2g35430 behavior during stress adaptation.

What considerations are important when developing immunohistochemistry protocols with At2g35430 antibody?

Developing robust immunohistochemistry (IHC) protocols for At2g35430 requires careful optimization of multiple parameters. Begin with fixation method selection; for plant tissues, 4% paraformaldehyde typically preserves antigen structure while maintaining tissue morphology. Test both paraffin embedding and cryosectioning approaches, as some antigens are better preserved in frozen sections. Section thickness (5-20 μm) affects antibody penetration and signal intensity, requiring empirical optimization.

Antigen retrieval methods are often critical for successful IHC; compare heat-induced epitope retrieval (in citrate buffer, pH 6.0) with enzymatic retrieval (using proteinase K) to determine optimal conditions. For the At2g35430 polyclonal antibody , titrate concentrations between 1:100 and 1:1000 in combination with various incubation times (2 hours at room temperature versus overnight at 4°C) to identify the optimal signal-to-noise ratio.

Plant tissues present unique challenges due to autofluorescence from cell walls, chlorophyll, and secondary metabolites. Address these through specific blocking steps such as 0.1% sodium borohydride treatment or Sudan Black B (0.1-0.3%) incubation prior to antibody application. When developing protocols, include critical controls: (1) omission of primary antibody, (2) isotype control (non-specific rabbit IgG), (3) peptide competition control, and (4) positive control tissue known to express At2g35430. Document all parameters meticulously, including section thickness, antigen retrieval conditions, blocking procedure, antibody concentrations, and incubation times/temperatures to ensure reproducibility.

How might nanobody technology complement traditional At2g35430 antibody applications?

Nanobody technology represents a promising complement to traditional polyclonal antibodies like the At2g35430 antibody . Derived from camelid heavy-chain-only antibodies, nanobodies are significantly smaller (~15 kDa versus ~150 kDa for conventional antibodies) with unique advantages for certain applications. Their small size enables superior tissue penetration and access to epitopes in crowded cellular environments or protein complexes that might be inaccessible to conventional antibodies. This property could be particularly valuable for studying At2g35430 in intact plant tissues or when the protein is part of multiprotein complexes.

Recent advances in nanobody development, such as the Adaptive Multi-Epitope Targeting and Avidity-Enhanced (AMETA) platform , demonstrate how engineered nanobody constructs can achieve exceptional target specificity and affinity. Similar approaches could potentially be applied to develop At2g35430-specific nanobodies with enhanced properties. Notably, nanobodies show remarkable stability under harsh conditions (high temperature, extreme pH, detergents) that would denature conventional antibodies, potentially allowing At2g35430 detection under experimental conditions where traditional antibodies fail.

For live-cell imaging applications, nanobodies can be genetically encoded as intrabodies and fused to fluorescent proteins, enabling real-time visualization of At2g35430 dynamics in living plant cells without fixation artifacts. Similarly, nanobody-based proximity labeling could map the At2g35430 interactome with higher spatial resolution than conventional Co-IP approaches. While traditional antibodies like the rabbit polyclonal At2g35430 antibody remain valuable for established techniques like Western blotting and ELISA, complementary nanobody development could expand the toolbox for studying this protein across multiple experimental contexts.

What role might At2g35430 antibody play in plant-pathogen interaction studies?

The At2g35430 antibody can serve as a valuable tool for investigating plant-pathogen interactions, particularly if the protein plays a role in plant immune responses or is targeted by pathogen effectors. In pathogen challenge experiments, Western blot analysis using the At2g35430 antibody can track protein abundance changes during infection, potentially revealing patterns of induction or suppression that suggest functional involvement in defense responses. Combining this with subcellular fractionation can identify pathogen-induced relocalization events, such as nuclear translocation that might indicate involvement in defense gene regulation.

Immunoprecipitation with the At2g35430 antibody following pathogen challenge can identify pathogen-specific interaction partners, potentially including pathogen effectors that target this protein. Such findings would suggest At2g35430 is a component of plant immunity. For plant-microbe interaction studies, immunohistochemistry using the At2g35430 antibody can visualize protein localization at infection sites, particularly at the plant-pathogen interface, providing spatial context for functional hypotheses.

When used in comparative studies across resistant and susceptible plant genotypes, the antibody can help determine whether At2g35430 abundance or modifications correlate with resistance phenotypes. For systems-level understanding, combine antibody-based protein detection with transcriptomics and metabolomics during infection to place At2g35430 within broader defense response networks. These approaches collectively can establish whether At2g35430 functions as a pattern recognition receptor, signaling component, transcriptional regulator, or direct antimicrobial protein in plant defense systems.

How can advances in antibody technologies enhance At2g35430 detection sensitivity and specificity?

Emerging antibody technologies offer significant opportunities to enhance At2g35430 detection. Single-molecule array (Simoa) technology, which compartmentalizes individual immunocomplexes in femtoliter-sized wells, could theoretically improve At2g35430 detection sensitivity by 100-1000 fold compared to conventional ELISA. This would enable detection of extremely low protein levels, particularly valuable for studying proteins with developmental stage-specific or stress-induced expression patterns.

Recent developments in recombinant antibody engineering could allow modification of the existing polyclonal At2g35430 antibody into more defined reagents. For example, identifying and cloning the highest-affinity antibody sequences from the polyclonal mixture could generate monoclonal antibodies with defined epitope specificities. These could then be engineered for specific applications through format modifications (Fab, scFv) or fusion to detection tags.

Bi-specific antibody formats, which simultaneously recognize two different epitopes, could enhance specificity by requiring recognition of two distinct regions of At2g35430 for signal generation. This approach could be particularly valuable in complex plant extracts where cross-reactivity with related proteins may occur. Similarly, proximity-dependent detection systems (like proximity ligation assays) can improve specificity by requiring two separate antibodies to bind in close proximity, essentially creating an AND gate for signal generation.

Integration with mass spectrometry through techniques like mass cytometry or immuno-MALDI could combine the specificity of antibody recognition with the precision of mass detection, potentially distinguishing between At2g35430 isoforms or post-translationally modified variants. As these technologies mature, they offer exciting possibilities for more sensitive, specific, and information-rich analysis of plant proteins like At2g35430.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.